(guselkumab)
This information is intended for US healthcare professionals to access current scientific information about J&J Innovative Medicine products. It is prepared by Medical Information and is not intended for promotional purposes, nor to provide medical advice.
Last Updated: 01/06/2025
Guselkumab is a human IgG1λ monoclonal antibody (mAb) that binds selectively to the IL-23) protein with high specificity and affinity. IL-23, a regulatory cytokine, affects the differentiation, expansion, and survival of T cell subsets, (eg, T helper 17 [Th17] cells and Tc17 cells) and innate immune cell subsets, which represent sources of effector cytokines, including IL-17A, IL-17F and IL-22 that drive inflammatory disease. In humans, selective blockade of IL-23 was shown to normalize production of these cytokines.1
Levels of IL-23 are elevated in the skin of patients with plaque PsO. In in vitro models, guselkumab was shown to inhibit the bioactivity of IL-23 by blocking its interaction with cell surface IL-23 receptor, disrupting IL-23-mediated signaling, activation and cytokine cascades. Guselkumab exerts clinical effects in plaque PsO and PsA through blockade of the IL-23 cytokine pathway.1
In a Phase 1 study, treatment with guselkumab resulted in reduced expression of IL-23/Th17 pathway genes and PsO-associated gene expression profiles, as shown by analyses of messenger RNA (mRNA) obtained from lesional skin biopsies of psoriatic subjects at week 12 compared to baseline. In the same Phase 1 study, treatment with guselkumab resulted in improvement of histological measures of PsO at week 12, including reductions in epidermal thickness and T-cell density. In addition, reduced serum IL-17A,
IL-17F and IL-22 levels compared to placebo were observed in guselkumab-treated subjects in Phase 2 and Phase 3 studies in plaque PsO. These results are consistent with the clinical benefit observed with guselkumab treatment in plaque PsO.1
In Phase 3 studies in PsA, evaluated subjects had elevated serum levels of acute phase proteins CRP, serum amyloid A and IL-6, and Th17 effector cytokines IL-17A, IL-17F and IL-22 at baseline. Guselkumab decreased levels of these proteins within 4 weeks of initiation of treatment. By week 24, guselkumab further reduced the levels of these proteins compared to baseline and also to placebo. In guselkumab-treated subjects, serum IL-17A and IL-17F levels were similar to those observed in a demographically matched healthy cohort at Week 24.1
Following a single 100 mg SC injection in healthy subjects, guselkumab reached a mean (± standard deviation; SD) Cmax of 8.09 ± 3.68 mcg/mL by approximately 5.5 days post dose.1
Steady-state serum guselkumab concentrations were achieved by Week 20 following SC administrations of 100 mg guselkumab at Weeks 0 and 4, and q8w thereafter. The mean (± SD) steady-state guselkumab Ctrough values in two Phase 3 studies in plaque PsO were 1.15 ± 0.73 mcg/mL and 1.23 ± 0.84 mcg/mL. Serum guselkumab concentrations did not appear to accumulate over time when given SC q8w.1
The PK of guselkumab in subjects with PsA was similar to that in subjects with plaque PsO. Following SC administration of 100 mg of guselkumab at Weeks 0, 4, and q8w thereafter, mean steady-state guselkumab Ctrough was approximately 1.2 mcg/mL. Following SC administration of 100 mg of guselkumab every 4 weeks (q4w), mean steady-state guselkumab Ctrough was approximately 3.8 mcg/mL.1
The absolute bioavailability of guselkumab following a single 100 mg SC injection was estimated to be approximately 49% in healthy subjects.1
Mean volume of distribution during the terminal phase (Vz) following a single IV administration to healthy subjects ranged from approximately 7-10 L (98-123 mL/kg) across studies.1
The exact pathway through which guselkumab is metabolized has not been characterized. As a human IgG mAb, guselkumab is expected to be degraded into small peptides and amino acids via catabolic pathways in the same manner as endogenous IgG.1
Mean systemic CL following a single IV administration to healthy subjects ranged from 0.288 to 0.479 L/day (3.6-6.0 mL/day/kg) across studies.1
Mean T1/2 of guselkumab was approximately 17 days in healthy subjects and approximately 15-18 days in subjects with plaque PsO across studies.1
The systemic exposure of guselkumab (Cmax and AUC) increased in an approximately dose-proportional manner following a single SC injection at doses ranging from 10 mg to 300 mg in healthy subjects or subjects with plaque PsO.1
In a population PK analysis, the apparent clearance (CL/F) and apparent volume of distribution (V/F) were 0.516 L/d and 13.5 L, respectively, and the T1/2 was approximately 18 days in subjects with PsO.1
In the population PK analysis, the effects of baseline demographics (weight, age, sex, and race), immunogenicity, baseline disease characteristics, comorbidities (past and current history of diabetes, hypertension, and hyperlipidemia), past use of therapeutic biologics, past use of methotrexate or cyclosporine, concomitant medications (nonsteroidal anti-inflammatory drug [NSAIDs], corticosteroids and conventional synthetic disease-modifying antirheumatic drugs [DMARDs] such as methotrexate), use of alcohol, or current smoking status, on PK of guselkumab was evaluated. Only the effects of body weight on CL/F and V/F were found to be significant, with a trend towards higher CL/F in heavier subjects. However, subsequent exposure-response modeling analysis suggested that no dose adjustment would be warranted for body weight.1
An in vitro study using human hepatocytes showed that IL-23 did not alter the expression or activity of multiple cytochrome P450 (CYP450) enzymes (CYP1A2, 2B6, 2C9, 2C19, 2D6, or 3A4).1
The effects of guselkumab on the PK of representative probe substrates of CYP isozymes (midazolam [CYP3A4], warfarin [CYP2C9], omeprazole [CYP2C19], dextromethorphan [CYP2D6], and caffeine [CYP1A2]) were evaluated in subjects with moderate to severe plaque PsO. Results from this study indicate that changes in Cmax and AUCinf of midazolam, S-warfarin, omeprazole, dextromethorphan, and caffeine after a single dose of guselkumab were not clinically relevant.1
There is no need for dose adjustment when co-administering guselkumab and CYP450 substrates.1
Zhuang et al (2016)2 conducted a phase 1, 2-part, randomized, double-blind, placebo-controlled, first-in-human, single-ascending-dose study of TREMFYA.
Chen et al (2022)3 conducted population PK modeling analyses and exposure-response modeling analyses to describe the PK of SC TREMFYA, quantify the effects of intrinsic or extrinsic factors that affect the PK variability, and characterize the relationship between guselkumab exposure and clinical efficacy measures in adult patients with active PsA receiving SC TREMFYA therapy.
Final Model
Simulations
Landmark Analyses
Longitudinal Modeling
Simulations
Probability of Achieving ACR Response | ACR20 | ACR50 | ACR70 | |||||
---|---|---|---|---|---|---|---|---|
TREMFYA 100 mg | TREMFYA 100 mg | TREMFYA 100 mg | ||||||
q4w | q8w | q4w | q8w | q4w | q8w | |||
DAS28 score | ||||||||
DAS28 ≤5.1 | 0.69 | 0.64 | 0.41 | 0.36 | 0.21 | 0.18 | ||
DAS28 >5.1 | 0.61 | 0.57 | 0.32 | 0.28 | 0.15 | 0.13 | ||
PASI score | ||||||||
PASI ≤5.8 | 0.62 | 0.57 | 0.33 | 0.29 | 0.15 | 0.13 | ||
PASI >5.8 | 0.69 | 0.64 | 0.40 | 0.35 | 0.20 | 0.17 | ||
Probability of Achieving IGA Response | IGA 0 | IGA 0/1 | ||||||
TREMFYA 100 mg | TREMFYA 100 mg | |||||||
q4w | q8w | q4w | q8w | |||||
PASI score | ||||||||
PASI ≤5.8 | 0.64 | 0.57 | 0.92 | 0.90 | ||||
PASI >5.8 | 0.53 | 0.44 | 0.88 | 0.84 | ||||
Abbreviations: ACR, American College of Rheumatology; ACR20/50/70, 20%/50%/70% improvement in the ACR response criteria from baseline; DAS28, Disease Activity Score in 28 joints; IGA 0, Investigator’s Global Assessment score of cleared (0); IGA 0/1, Investigator’s Global Assessment score of cleared (0) or minimal (1); PASI, Psoriasis Area and Severity Index; q4w, every 4 weeks; q8w, every 8 weeks. |
Tran et al (2022)4 described the PK of TREMFYA across 9 clinical studies, including a phase 1 study in healthy patients and multiple phase 2 and 3 studies in different patient populations (PsO, PsA, and PPP) using a population PK modeling approach. Covariates that explained intersubject variability were identified and relationships between covariates were assessed.
Study | Study Design | Population | TREMFYA Treatment Schedule (Duration) | Sampling Time Points |
---|---|---|---|---|
Phase 3, multicenter, randomized, double-blind studies | ||||
PSO3001 (VOYAGE-1) | Placebo and active comparator-controlled study | Patients with moderate to severe plaque-type PsO | SC TREMFYA 100 mg at wks 0 and 4, then q8w (48 wks) | Wk 0, 4, 8, 12, 16, 20, 24, 28, 36, 44 Random: wk 16-24 |
PSO3002 (VOYAGE-2) | Placebo and active comparator-controlled study | Patients with moderate to severe plaque-type PsO with randomized withdrawal and retreatment | SC TREMFYA 100 mg at wks 0 and 4, then q8w (72 wks) | Wk 0, 4, 8, 12, 16, 20, 24, 28, 32, 36, 40, 44 |
PSA3001a | Placebo-controlled study | Patients with active PsA, including those previously treated with ≥1 anti-TNF alpha agent | SC TREMFYA 100 mg q4w SC TREMFYA 100 mg at wks 0 and 4, then q8w (48 wks) | Wk 0, 4, 8, 12, 16, 20, 24, 28, 36, 44, 52, 60 Random: wk 4-12 |
PSA3002a (DISCOVER-2) | Placebo-controlled study | Patients with active PsA | SC TREMFYA 100 mg q4w SC TREMFYA 100 mg at wks 0 and 4, then q8w (100 wks) | Wk 0, 2, 4, 8, 12, 16, 20, 24, 28, 36, 44, 52, 76, 100 Random: wk 4-12 |
PPP3001 | Placebo-controlled study | Patients with palmoplantar pustulosis | SC TREMFYA 100 or 200 mg at wks 0 and 4, then q8w (60 wks) | Wk 0, 4, 8, 12, 16, 20, 24, 28, 36, 44, 48, 52, 60 Random: wk 16-20 |
Phase 2, randomized, double-blind studies | ||||
PSO2001 (X-PLORE) | Placebo- and active comparator-controlled, dose-ranging study | Patients with moderate to severe plaque-type PsO | SC TREMFYA 5, 50, and 200 mg at wks 0 and 4, then q12w SC TREMFYA 15 and 100 mg q8w (40 wks) | Wk 0, 4, 8, 12, 16, 20, 24, 28, 32, 36, 40 |
PSA2001a | Phase 2a, multicenter, placebo-controlled study | Patients with active PsA | SC TREMFYA 100 mg at wks 0 and 4, then q8w (56 wks) | Wk 0, 4, 8, 12, 16, 20, 24, 28, 36, 44, 56 Random: wk 16-24 |
PPP2001 | Multicenter, placebo-controlled study | Patients with palmoplantar pustulosis | SC TREMFYA 200 mg at wks 0 and 4 (24 wks) | Wk 0, 1, 2, 4, 6, 8, 12, 16, 20, 24 |
Phase 1, randomized study | ||||
NAP1001 | Open label, parallel study to assess the PK comparability of 2 formulations and 2 different devices | Healthy subjects | Single SC TREMFYA 100 mg injection (lyophilized formulation, PFS-U or PFS-FID) Single IV TREMFYA 100 mg infusion | D 1 (predose, 1 h, 4 h, 12 h), D 2, 3, 4, 5, 6, 7, 15, 22, 29, 43, 57, 71, 85 |
Abbreviations: D, day; h, hour; IV, intravenous; NONMEM, nonlinear mixed-effects modeling; PFS-FID, prefilled syringe facilitated injection device; PFS-U, UltraSafe Passive™ Delivery System; PK, pharmacokinetic; PsA, psoriatic arthritis; PsO, psoriasis; q4w, every 4 weeks; q8w, every 8 weeks; q12w, every 12 weeks; SC, subcutaneous; TNF, tumor necrosis factor; wk, week. aAnalysis includes data up to wk 24 (due to data cut-off date for NONMEM file creation). |
Covariates | Effect of Covariates, Change Relative to Reference (90% CI) | |
---|---|---|
Effect on CL | ||
Body weight (97/70 kg) | 1.37 (1.31-1.44) | |
PsA (yes/no) | 0.85 (0.82-0.88) | |
Plaque PsO (yes/no) | 0.81 (0.78-0.84) | |
PPP (yes/no) | 0.82 (0.78-0.86) | |
Non-white race (yes/no) | 1.15 (1.12-1.18) | |
Diabetes (yes/no) | 1.14 (1.11-1.18) | |
Very high CRP (>10 mg/dL) | 1.07 (1.03-1.11) | |
Albumin (47/43 g/L) | 0.93 (0.92-0.95) | |
High CRP (>3 mg/dL) | 1.05 (1.03-1.07) | |
Current smoker (yes/no) | 1.06 (1.04-1.08) | |
Sex (female/male) | 1.05 (1.03-1.07) | |
White blood cell count (8.2/5.7x109 | 1.05 (1.04-1.06) | |
NSAID use (yes/no) | 0.96 (0.94-0.97) | |
ALP (89/62 U/L) | 1.02 (1.01-1.03) | |
Effect on Vc | ||
Body weight (97/70 kg) | 1.48 (1.4-1.57) | |
Abbreviations: ALP, alkaline phosphatase; CI, confidence interval; CL, clearance; CRP, C-reactive protein; NSAID, nonsteroidal anti-inflammatory drug; PK, pharmacokinetic; PPP, palmoplantar pustulosis; PsA, psoriatic arthritis; PsO, psoriasis; Vc, central volume of distribution. |
A literature search of MEDLINE®
1 | Data on File. Guselkumab. Company Core Data Sheet. Janssen Research & Development, LLC. EDMS-ERI-111962822; 2024. |
2 | |
3 | |
4 |